Weekly Digests
‹ Back to November

MiTEs turn TAM suppression into T cell synergy

November 26, 2025

Inhibition or reprogramming of tumor-associated macrophages (TAMs) has great potential to improve the tumor immune environment, but has, so far, shown limited clinical efficacy. To improve macrophage-targeted therapies, von Locquenghien, Zwicky, Cie, et al. described their dual TAM- and T cell-targeting strategy in a recent Cell publication.

Using publicly available datasets from human lung, breast, colon, and ovarian tumors, the researchers started by characterizing the spatial interactions between TAMs and other immune cells in the tumor microenvironment (TME). TAMs were the main population observed near T cells. Next, various algorithms were applied to a single-cell RNA sequencing atlas with human tumor samples and adjacent healthy tissues to assess cell–cell communication. This also showed that the predominant interaction was between TAMs and T cells, and involved chemokines related to T cell recruitment, costimulatory and inhibitory interactions, and immunosuppressive signals, which were all upregulated in the TME.

Based on these data, the researchers developed a treatment strategy to simultaneously reprogram TAMs and activate T cells in the TME. Previous work revealed TREM2 as an important inhibitory receptor on TAMs. Therefore, an antagonistic anti-human TREM2 antibody was developed and tested in humanized TREM2 mice. Treatment with this antibody prevented the induction of the TAM program, and preserved pro-inflammatory characteristics in vitro in bone-marrow-derived macrophages. However, the treatment had limited efficacy in mice with MC38 tumors. To improve efficacy, a myeloid-targeting immunocytokine (ICK) was created by fusing the anti-TREM2 antibody to IL-2, enabling it to act on both TAMs and T cells in the TME. A synthetic IL-2 variant (superkine [SK]) with increased affinity for IL-2Rβ was used, as it preferentially targets effector T cells over Tregs. However, intraperitoneal treatment of MC38-bearing mice with anti-TREM2-IL-2SK resulted in death, caused by systemic toxicity and hepatotoxicity.

To overcome this systemic toxicity, the researchers developed an inactive IL-2SK domain (pro-cytokine) containing a blocking domain (IL2-Rβ) with a protease-cleavable linker, which could be activated locally by TAM-associated proteases. To determine which protease to use for this construct, scRNAseq analysis of protease expression was performed on immune cells from a human cancer atlas, and PBMCs from healthy donors. TAMs showed higher expression of several proteases, and MMP14 was selected for its high expression in TAMs and limited expression in other immune cells. There was also a strong correlation between MMP14 and TREM2 expression in both human and murine tumors. Based on this, four variants of myeloid-targeted ICKs (MiTEs) were designed, and one of these was selected for further assessment. MiTE144 showed high affinity and specificity for TREM2, did not bind IL-2Rβ in its intact form, induced strong T cell proliferation after protease treatment, and had limited impact on Treg activity.

To determine its toxicity, mice inoculated with MC38 cells received MiTE144 intravenously every 3 days starting 10 days after tumor inoculation. No systemic inflammation or hepatotoxicity was observed. For efficacy analysis, mice were treated on days 8, 10, and 13 after MC38 tumor inoculation. MiTE144 monotherapy significantly reduced tumor growth and was more effective than anti-TREM2 monotherapy, immune checkpoint blockade (ICB) monotherapies, and combinations of anti-CTLA-4 and anti-PD-1 with anti-TREM2. The combination of MiTE144 with anti-CTLA-4 further improved its efficacy, with complete tumor eradication in 6/7 mice.

The researchers then analyzed the effects of MiTE144 on tumoral immune populations using scRNAseq on day 11 post-tumor inoculation, after mice received treatment on days 8 and 10. A deep generative model was used to construct treatment-specific distance similarity networks. Treatments with MiTE144, +/- anti-PD-1/CTLA-4, formed a distinct cluster. Differential expression analysis showed that therapies with MiTE144 triggered the most extensive transcriptional reprogramming in macrophages, DCs, T cells, and NK cells. It reduced IFN-responsive TAMs and DCs while increasing hypoxia-associated TAMs and granulocytes. In contrast, it increased CD45+ immune cell counts, monocytes, and granulocytes in TDLNs.

Using a deep learning model to assess the effect of each treatment relative to other treatments revealed that MiTE144 upregulated inflammatory monocyte-like and hypoxia-associated modules, while reducing IFN-response genes and modules. It also downregulated the suppressive hTREM2/Mmp14 modules in macrophages, while upregulating Trem1/3, suggesting a shift toward earlier, inflammatory monocytic states.

MiTE144 alone or combined with anti-PD-1/CTLA-4 increased NK cell proliferation, resulting in higher NK cell counts in the TME and TDLN. NK cells upregulated late/activation, cytotoxic, and proliferation genes, while early exhaustion and dysfunction-related genes were downregulated, suggestive of a shift toward a highly proliferative and cytotoxic phenotype.

Treg frequencies increased slightly in the TME and more in the TDLN, but this effect was not present when treatment was combined with anti-CTLA-4. CD8+ T cells increased in both the TME and TDLN in response to MiTE144, and these cells upregulated cytotoxic, proliferation, and stemness-associated genes, while exhaustion-associated genes were downregulated. These effects were more pronounced when MiTE was combined with anti-CTLA-4.

To determine whether these effects could also be observed in human tumors, patient-derived tumor fragments (PDTFs) obtained from five patients with renal cell carcinoma were treated ex vivo with MiTE144 and/or anti-PD-1, and immune cells were analyzed by CITEseq after 48 hours. One non-responding PDTF was excluded from analysis. In the other PDTFs, MiTE144 +/- anti-PD-1 resulted in shifts in immune cell populations, with expansion of CD8+ cycling and memory T cells and intermediate-stage NK cells. Tregs and hypoxic TAMs remained unchanged or reduced compared to controls. T cells upregulated cytolytic effector genes and cell cycle-associated genes, while markers associated with exhaustion decreased. In NK cells, treatment also induced cytotoxic programs, proliferation-associated genes, and activation markers, while downregulating exhaustion genes.

Overall, this MiTE strategy enabled reprogramming of TAMs, improved T cell activity, and worked synergistically with ICB. If shown to be effective and safe in patients, it may serve as an interesting new combinatory strategy with ICB, particularly for tumor types with high numbers of immunosuppressive TAMs that currently show limited response to ICB treatment.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, co-first author Michelle von Locquenghien answered our questions.

On the left, co-first author Michelle von Locquenghien; on the right the Amit lab.

What was the most surprising finding of this study for you?
The most unexpected result emerged during the first generation of anti-TREM2–IL-2 constructs. We initially assumed that the TREM2-binding Fab would provide sufficient tumor selectivity to prevent off-target cytokine activity. Instead, our first in vivo studies in transgenic hTREM2 mice resulted in acute cytokine-driven systemic toxicity, revealing how deceptively small design assumptions can have major biological consequences. These results prompted a complete reassessment of our engineering strategy, and reshaped our underlying hypothesis. In parallel, while we anticipated strong T cell activation and proliferation in response to the IL-2 stimuli, the magnitude and consistency of NK cell cytotoxicity and proliferation across models were far greater than expected. This synergistic myeloid–lymphoid response may represent a critical advantage for overcoming challenges that limit T cell-centric immunotherapies, such as MHC-I loss.

What is the outlook?
This work positions MiTEs as a next-generation macrophage-targeted immunocytokine platform that integrates conditional cytokine activation with immune checkpoint blockade. Because MiTEs function independently of tumor antigens and become active specifically within the tumor microenvironment, they offer broad applicability across solid tumors while avoiding systemic toxicity, overcoming key limitations of traditional cytokine therapies. MiTEs show promising results in human patient-derived tumor fragments and synergize strongly with existing immune checkpoint inhibitors. The fully humanized design supports a clear translational path, including non-human primate studies and eventual clinical evaluation. Beyond IL-2, the platform can be extended to additional cytokines and immune axes to overcome resistance mechanisms and drive multi-dimensional immune reprogramming in cancer therapy.

If you could go back in time and give your early-career self one piece of advice for navigating a scientific career, what would it be?
View unexpected results as strategic opportunities rather than setbacks; some of the most impactful insights arise when data challenge your assumptions. And be willing to pursue ambitious, high-risk ideas. Transformative research often begins with daring to explore concepts that push past traditional approaches.

References:

von Locquenghien M, Zwicky P, Xie K, Jaitin DA, Sheban F, Yalin A, Uhlitz F, Gur C, Eshed RS, David E, Mazuz K, Marin CJ, Sankar A, Mediratta D, Avellino R, Weiner A, Amit I. Macrophage-targeted immunocytokine leverages myeloid, T, and NK cell synergy for cancer immunotherapy. Cell. 2025 Nov 19.

In the Spotlight...

A modular mRNA platform for programmable induction of tumor-specific immunogenic cell death

Dong et al. encoded the ICD-inducing protein 4HB in LNPs optimized for tumor cell specificity and hepatocyte avoidance, which significantly improved tumor cargo expression compared to a clinical control LNP (>90% vs. ~25%). mRNA engineering via inclusion of UTRs complementary to non-tumor miRNAs (suppressive for liver) reduced off-target cargo distribution, markers of tissue injury, and body weight loss, without compromising tumor expression. The platform synergized with ICB in diverse “cold” tumor models via i.t. and i.v. routes, without systemic toxicity, enhancing infiltration of diverse immune cell types, including CD8+ T cells and macrophages.

Contributed by Morgan Janes

CD8+ T cell antitumor immunity via human iNKT-DC conjugates

Baiu et al. showed that CD4+ invariant natural killer T cells (iNKT) and autologous or allogeneic monocyte-derived DCs formed stable complexes, with enhanced expression of MHC I, 4-1BBL, OX40L, and IL-15Ra in DCs, and CD70 in iNKT cells. Complexes generated sustained DC signaling and created a platform for antigen-specific CD8+ T cell activation. In a xenograft model of B cell lymphoma, iNKT-DC induced T cell effector differentiation, reduced tumor burden, and remained effective at late disease stages that were resistant to ICB. Patient-derived DCs formed similar conjugates with allogeneic CD4+ iNKT cells and activated tumor antigen-specific CD8+ T cells.

Contributed by Shishir Pant

IL-9 as a naturally orthogonal cytokine with optimal JAK/STAT signaling for engineered T cell therapy

Taking advantage of low IL-9R expression in normal tissues, Jiang et al. engineered T cells with wild-type IL-9R. After transfer into tumor-bearing mice in combination with IL-9, which was well tolerated, IL-9R-engineered T cells showed enhanced infiltration, stemness, and antitumor activity compared to similar studies with orthogonal IL-9R. The IL-9–IL-9R axis activated STAT1/3/5 and (unexpectedly) recruited STAT4, and was highly sensitive to alterations in IL-9R signal strength or pSTAT stoichiometry. Levels of pSTAT1 determined differentiation towards stem/memory or terminal effector states. Similar results were observed CAR T cells.

Contributed by Lauren Hitchings

Intratumoral three-cell-type clusters are a conserved feature of endogenous antitumor immunity

Damle and Carter et al. integrated spatial proteomics, tissue microarray, multiplex immunofluorescence imaging, and transcriptomics data from highly desmoplastic fibrolamellar carcinoma, and identified DC:Th:CTL three-cell-type clusters were present non-randomly in the TME. Immune triads were enriched for cDC1, mregDC, CXCL13+ Th, and GZMK+ effector CTL cells, showed upregulation of immune activation markers, and were present in treatment-naive murine and human PDAC. The density of APC:Th:CTL three-cell-type clusters in primary PDAC samples (n=467) correlated with intratumoral clonal T cell expansion and improved OS.

Contributed by Shishir Pant

Ms4a7 expression in cDC1s determines cross-­presentation and antitumor immunity

Focused on key TME regulators that control cDC1 cross-presentation, Xie et al. found that cDC1-expressed Ms4a7 was critical for effective CD8+ T cell-mediated antitumor immunity in mouse models. Tumor antigen induced Ms4a7 expression and NF-κB activation in cDC1s and tumor dLNs, which was required to cross-prime antigen-specific CD8+ T cells. Ms4a7-/- cDC1s showed reduced migration to dLNs, made less IL-­12, IL-­18, and IL-­27 in the TME, and failed to prime CD8+ T cell activation, resulting in diminished antitumor immunity. In human cancers, Ms4a7 was expressed in a subset of cDC1s, enriched in dLNs, and correlated with patient survival.

Contributed by Katherine Turner

Predominant mutated non-canonical tumor-specific antigens identified by proteogenomics demonstrate immunogenicity and tumor suppression in CRC

Xiang et al. integrated whole-genome, transcriptomics, and MHC-I immunopeptidomics analyses to identify tumor-specific antigens from non-coding regions in colorectal cancer samples. Across 10 paired samples, over 80% of 96 MHC-I-presented neo-epitopes originated from intergenic and intronic regions. Hypermutated tumors showed the highest burden of non-canonical neo-epitopes, while non-hypermutated tumors relied on coding alterations and alternative splicing. In the subcutaneous MC38 model, multi-epitope vaccines containing mutated non-canonical neo-epitopes effectively activated CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner.

Contributed by Shishir Pant

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.